GB2553170A - Promoter - Google Patents

Promoter Download PDF

Info

Publication number
GB2553170A
GB2553170A GB1703224.4A GB201703224A GB2553170A GB 2553170 A GB2553170 A GB 2553170A GB 201703224 A GB201703224 A GB 201703224A GB 2553170 A GB2553170 A GB 2553170A
Authority
GB
United Kingdom
Prior art keywords
seq
polynucleotide
nucleic acid
acid molecule
nucleotide sequence
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
GB1703224.4A
Other versions
GB201703224D0 (en
GB2553170B (en
Inventor
Cawood Ryan
Parker Manuel Richard
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Oxford Genetics Ltd
Original Assignee
Oxford Genetics Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Oxford Genetics Ltd filed Critical Oxford Genetics Ltd
Priority to GB2002193.7A priority Critical patent/GB2578859B/en
Publication of GB201703224D0 publication Critical patent/GB201703224D0/en
Publication of GB2553170A publication Critical patent/GB2553170A/en
Application granted granted Critical
Publication of GB2553170B publication Critical patent/GB2553170B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • C07H21/04Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids with deoxyribosyl as saccharide radical
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/001Vector systems having a special element relevant for transcription controllable enhancer/promoter combination
    • C12N2830/002Vector systems having a special element relevant for transcription controllable enhancer/promoter combination inducible enhancer/promoter combination, e.g. hypoxia, iron, transcription factor
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/80Vector systems having a special element relevant for transcription from vertebrates
    • C12N2830/85Vector systems having a special element relevant for transcription from vertebrates mammalian

Landscapes

  • Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Microbiology (AREA)
  • Medicinal Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Virology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Peptides Or Proteins (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

An enhanced promoter for use in an expression vector in mammalian cells comprises a core promoter sequence SEQ ID NO:4 (or a sequence having 80% or more sequence identity) with one of SEQ ID 1, 2 or 3 (or a sequence having 80% or more sequence identity) operably positioned 5 and one of SEQ ID NO5, 6 or 7 (or a sequence having 80% or more sequence identity) operably positioned 3 (See Table 1 at page 17). Preferably the sequences are contiguous as exemplified and are stronger or have greater activity than a standard CMV promoter. Vectors, cells and kits comprising the promoters are disclosed.

Description

(54) Title of the Invention: Promoter
Abstract Title: Enhanced promoter for heterologous expression in mammalian cells (57) An enhanced promoter for use in an expression vector in mammalian cells comprises a core promoter sequence SEQ ID NO:4 (or a sequence having 80% or more sequence identity) with one of SEQ ID 1,2 or 3 (or a sequence having 80% or more sequence identity) operably positioned 5’ and one of SEQ ID NO5, 6 or 7 (or a sequence having 80% or more sequence identity) operably positioned 3’ (See Table 1 at page 17). Preferably the sequences are contiguous as exemplified and are ‘stronger’ or have greater activity than a standard CMV promoter. Vectors, cells and kits comprising the promoters are disclosed.
At least one drawing originally filed was informal and the print reproduced here is taken from a later filed formal copy.
- 1 / 10 Figure 1 ./ί&/ II
/ΜΙΠ mri
- 2 / 10 Figure 2
3/10
r-.
ο © ©
4*
ί 8 j S i 8 yj
X * χ·
-4/10Figure 4 tsj
0)
4·* o
£
ΙΛ
CL >
s o
«0
ΙΛ • w • «_ • * · • t»
MMB
MM '3
Imm
Q.
>
£
O
I!1S ··«
X c
o cu u>
1/
O
ϋβ
ΙΛ
M o
ex >
£
Q
- 5 / 10 -
w
- 6 / 10 -
a. ¢+
Λχ ·' %
\£ί
O-
lx t<V
Ί.
% : '$+
- 7 / 10 -
- 8 / 10 -
Ό·· ::04
X»*·
NS
- 9 / 10 Figure 9
>
δ ¢0 t;
8.Ϊ
Φ
4(X
M...
(5 ί£ί
7y
Q
Φ ΐΗ
G
X
\O
ΓΧ-.
W'J sx
;.r>
CX
R R 8 8 ρ£ \0 $ V &
L-W
- 10/10-
. i .
PROMOTER
The present invention relates to nucleic acid molecules which are capable of promoting transcription of operably-linked heterologous polynucleotides in mammalian cells. The invention also relates to expression vectors and host ceils which comprise the nucleic acid molecules of the invention. Such expression vectors may be used to produce recombinant proteins, e.g. antibodies and lentiviral polypeptides.
For transcription of a gene to occur, RNA polymerase must bind to the gene promoter and initiate transcription. In general, RNA polymerase I transcribes genes encoding ribosomal RNA; RNA polymerase II transcribes genes encoding messenger RNA, some small nuclear RNAs and microRNAs; while RNA polymerase ill transcribes genes encoding transfer RNAs and other small RNAs.
Transcription is regulated in order to control when transcription occurs and how much RNA is created. Transcription of a gene by RNA polymerase can be regulated by at least five mechanisms:
(i) Specificity factors alter the specificity of RNA polymerase for a given promoter or set of promoters, making it more or less likely to bind to them (e.g. sigma factors used in prokaryotic transcription).
(ii) Repressors bind to the Operator (coding sequences on the DNA strand that are close to or overlapping the promoter region) impeding RNA polymerase's progress along the strand, thus impeding the expression of the gene.
(iii) Transcription factors position RNA polymerase at the start of a protein-coding sequence and then release the polymerase to transcribe the mRNA.
(iv) Activators enhance the interaction between RNA polymerase and a particular promoter, encouraging the expression of the gene. Enhancers are sites on the DNA helix that are bound by activators in order to loop the DNA bringing a specific promoter to the initiation complex.
(v) Silencers are regions of DNA sequences that, when bound by particular transcription factors, can silence expression of the gene.
A typical mammalian promoter consists of a 50-100 base pair core region to which the basic transcription machinery binds, and an enhancer region to which one or more transcriptional activator proteins (transactivators) may bind. The number and type of transactivators that are able to bind at an enhancer depends on which specific binding sites are present. The rate of initiation is governed by the number and type of transactivators actually bound at a promoter’s enhancer. In addition to enhancers, there are silencers which, when bound by different transcription factors, lower gene expression.
Core promoters are made up of various different elements, of which there are two categories: canonical and non-canonical. Canonical core promoter elements include: TATA box, the initiator (Inr), the TFIIB recognition element (BRE), downstream promoter element (DPE) and downstream core element (DCE). These elements may be found within the core promoters of many but not all protein-coding genes. The TATA box (sequence TATAA) is usually found 20-30 bp upstream of the transcription start site (TSS) and acts as a binding site for the TFIID general transcription factor. When the Inr element (consensus sequence YYANT/AYY) is present, it encompasses the TSS, with the first A of the consensus being the first base of the transcript. BRE elements can be found both upstream of the TATA box (BREu consensus G/C G/C G/A CGCC) or downstream (BREd consensus G/A T T/A T/G T/G T/G T/G). Although not technically a canonical core promoter element, the CCAAT box (located between 50 and 100 bp upstream the TSS) is often included in this category. The CCAAT box also contributes to general transcription factor (TF) binding.
Non-canonical core promoter elements include the CpG island, the ATG desert and the transcription initiation platform (TIP). CpG islands generally span a 500-2000 bp stretch of DNA that contains a relatively high proportion of CpG dinucleotides. CpG dinucleotides would normally be methylated on the C residue, reducing transcription, but within CpG islands they remain unmethylated, promoting transcription. An ATG desert is a region of DNA with a lower frequency of ATG trinucleotides than surrounding regions. They extend approximately 1000 bp up and downstream of the TSS and are generally associated with promoters that do not contain TATA boxes.
- 3 A strong promoter is one that initiates transcription with a high frequency and can be a very useful tool. In biochemistry for example, strong promoters can be used to study transcription processes or to drive the production of recombinant proteins. Strong promoters are also useful in genetics: for example, they can be used to drive shRNA expression for gene knockdowns or for cDNA overexpression to deduce a protein's function.
A particularly potent promoter could also have medical applications: in a recombinant virus vaccine for example, higher antigen expression results in a better immune response.
The strongest promoters used In mammalian systems generally come from either constitutiveiy expressed cellular genes or from viral genes. However, the promoters which are the strongest in terms of the expression levels of an associated gene are often the most cell-type dependent ones, I.e. they are limited in the types of ceils in which they will work.
It is an object of the invention therefore to provide nucleic acid molecules which are capable of promoting transcription of an operably-linked heterologous polynucleotide at high levels and/or in a range of mammalian cells.
It is another object of the invention to provide expression vectors which comprise the nucleic acid molecules of the invention. Such expression vectors may be used to produce high levels of recombinant polypeptides, e.g. antibodies and lentiviral polypeptides.
In one embodiment, the invention provides a nucleic acid molecule comprising:
(a) a first polynucleotide having at least 80% nucleotide sequence identity to the nucleotide sequence set forth in SEQ ID NO: 1, SEQ ID NO: 2 or SEQ ID NO: 3, or a functional fragment of that first polynucleotide; and (b) a second polynucleotide having at least 80% nucleotide sequence identity to the nucleotide sequence set forth in SEQ ID NO: 4, or a functional fragment of that second polynucleotide; wherein (a) and (b) are joined 5’-3’ in this order, and wherein said nucleic acid molecule is capable of promoting transcription of an operably-linked heterologous polynucleotide in a mammalian cell.
-4In a further embodiment, the invention provides an expression vector comprising a nucleic acid molecule of the invention.
In yet a further embodiment, the invention provides an expression vector comprising the nucleic acid molecule of the invention, wherein the nucleic acid molecule is operably-linked to a heterologous polynucleotide.
The invention also provides a mammalian host ceil comprising an expression vector of the invention.
The invention also provides a kit comprising an expression vector of the invention.
In some embodiments, the nucleic acid molecule is an isolated nucleic acid molecule. The nucleic acid molecule of the invention is capable of promoting transcription of an operably-linked heterologous polynucleotide in a mammalian ceil, i.e. it is acting as a promoter. In some embodiments, the nucleic acid molecule of the invention acts as a constitutive promoter.
The nucleic acid molecule of the invention comprises (a) a first polynucleotide having at least 80% nucleotide sequence identity to the nucleotide sequence set forth in SEG ID NO: 1, SEQ ID NO: 2 or SEQ ID NO: 3. The sequences of SEG ID NOs: 1-3 are given in the attached “Sequences” section. The first polynucleotide preferably has at least 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% nucleotide sequence identity to one of the nucleotide sequences set forth in SEG ID NO: 1, SEQ ID NO: 2 or SEQ ID NO: 3. Most preferably, the first polynucleotide has the nucleotide sequence of SEQ ID NO: 1, SEQ ID NO: 2 or SEQ ID NO: 3.
The nucleic acid molecule of the invention may alternatively comprise a functional fragment of the first polynucleotide. As used herein, the term “functional fragment of that first polynucleotide” refers to a portion of the first polynucleotide that retains at least 20% (e.g. at least 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% or 100%) of the activity of the complete first polynucleotide in promoting the expression of an operably-linked heterologous polynucleotide. Methods for measuring and comparing the promoter activity of nucleic acid sequences are well known in the art, as discussed below.
Preferably, the functional fragment of the first polynucleotide is at least 50%, 60%, 70%, 80%, 90% or 95% of the length of the polynucleotide sequence set forth in SEQ ID NO: 1, SEG ID NO: 2 or SEQ ID NO: 3.
The nucleic acid molecule of the invention also comprises: (b) a second polynucleotide having at least 80% nucleotide sequence identity to the nucleotide sequence set forth in SEQ ID NO: 4, or a functional fragment of that second polynucleotide. The sequence of SEQ ID NO: 4 is given in the attached “Sequences” section. Preferably, the second polynucleotide has at least 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% nucleotide sequence identity to the polynucleotide sequence set forth in SEQ ID NO: 4. Most preferably, the second polynucleotide has the polynucleotide sequence of SEQ ID NO: 4.
The nucleic acid molecule of the invention may alternatively comprise a functional fragment of the second polynucleotide. As used herein, the term “functional fragment of that second polynucleotide” refers to a portion of the second polynucleotide that retains at least 20% (e.g. at least 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%,
90%, 95%, 98%, 99% or 100%) ofthe activity ofthe complete second polynucleotide in promoting the expression of an operably-linked heterologous polynucleotide. Methods for measuring and comparing the promoter activity of nucleic acid sequences are well known in the art, as discussed below. Preferably, the functional fragment of the second polynucleotide is at least 50%, 60%, 70%, 80%, 90% or 95% of the length of the polynucleotide sequence set forth in SEQ ID NO: 4.
SEQ ID NO: 4 comprises a Transcription Start Site (TSS): the +1 site (i.e. the first base to be transcribed) is the first T in the sequence TCAGATC; this occurs at the 3’end of SEQ ID NO: 4. Everything downstream of the TSS will be transcribed into RNA.
It is preferred, therefore, that the second polynucleotide, or the functional fragment thereof, comprises the sequence TCAGATC.
In some preferred embodiments, the nucleic acid molecule comprises:
(a) a first polynucleotide having the nucleotide sequence set forth in SEQ ID NO:
1, SEQ ID NO: 2 or SEQ ID NO: 3; and (b) a second polynucleotide having the nucleotide sequence set forth in SEQ ID
NO: 4;
wherein (a) and (b) are joined 5!-3’, and wherein said nucleic acid molecule is capable of promoting transcription of an operably-linked heterologous polynucleotide in a mammalian cell.
- 6 In some preferred embodiments, the nucleic acid molecule comprises:
(a) a first polynucleotide having the nucleotide sequence set forth in SEQ ID NO: 1; and (b) a second polynucleotide having the nucleotide sequence set forth in SEQ ID NO: 4;
wherein (a) and (b) are contiguously joined 5!-3’ in this order, and wherein said nucleic acid molecule is capable of promoting transcription of an operably-linked heterologous polynucleotide in a mammalian cell.
In some preferred embodiments, the nucleic acid molecule comprises:
(a) a first polynucleotide having the nucleotide sequence set forth in SEQ ID NO: 2; and (b) a second polynucleotide having the nucleotide sequence set forth in SEQ ID NO: 4;
wherein (a) and (b) are contiguously joined 5!-3’ in this order, and wherein said nucleic acid molecule is capable of promoting transcription of an operably-linked heterologous polynucleotide in a mammalian cell.
In some preferred embodiments, the nucleic acid molecule comprises:
(a) a first polynucleotide having the nucleotide sequence set forth in SEQ ID NO:
3; and (b) a second polynucleotide having the nucleotide sequence set forth in SEQ ID
NO: 4;
wherein (a) and (b) are contiguously joined 5!-3’ in this order, and wherein said nucleic acid molecule is capable of promoting transcription of an operably-linked heterologous polynucleotide in a mammalian cell.
The invention also provides a nucleic acid molecule comprising:
(a) a first polynucleotide having at least 80% nucleotide sequence identity to the nucleotide sequence set forth in SEQ ID NO: 1, SEQ ID NO: 2 or SEQ ID NO: 3, or a functional fragment of that first polynucleotide;
(b) a second polynucleotide having at least 80% nucleotide sequence identity to the nucleotide sequence set forth in SEQ ID NO: 4, or a functional fragment of that second polynucleotide; and >~ί ! “ (c) a third polynucleotide having at least 80% nucleotide sequence identity to the nucleotide sequence set forth in SEQ ID NO: 5, SEQ ID NO: 6 or SEQ ID NO: 7, or a functional fragment of that third polynucleotide;
wherein (a), (b) and (c) are joined 5’-3’ in this order, and wherein said nucleic acid molecule is capable of promoting transcription of an operably-linked heterologous polynucleotide in a mammalian cell.
The sequences of SEQ ID NOs: 5-7 are given in the attached “Sequences” section.
The third polynucleotide preferably has at least 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% nucleotide sequence identity to one of the nucleotide sequences set forth in SEQ ID NO: 5, SEQ ID NO: 6 or SEQ ID NO: 7. Most preferably, the third polynucleotide has the nucleotide sequence of SEQ ID NO: 5, SEQ ID NO: 6 or SEQ ID NO: 7.
The nucleic acid molecule of the invention may alternatively comprise a functional fragment of the third polynucleotide. As used herein, the term “functional fragment of that third polynucleotide” refers to a portion of the third polynucleotide that retains at least 20% (e.g. at least 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% or 100%) of the activity of the complete third polynucleotide in promoting the expression of an operably-linked heterologous polynucleotide. Methods for measuring and comparing the promoter activity of nucleic acid sequences are well known in the art, as discussed below.
Preferably, the functional fragment of the third polynucleotide is at least 50%, 60%, 70%, 80%, 90% or 95% of the length of the polynucleotide sequence set forth in SEQ ID NO: 5, SEQ ID NO: 6 or SEQ ID NO: 7.
In some preferred embodiments, the nucleic add molecule comprises:
(a) a first polynucleotide having the nucleotide sequence set forth in SEQ ID NO:
1, SEQ ID NO: 2 or SEQ ID NO: 3;
(b) a second polynucleotide having the nucleotide sequence set forth in SEQ ID
NO: 4; and .Βίο) a third polynucleotide having the nucleotide sequence set forth in SEG ID NO: 5, SEG ID NO: 6 or SEG ID NO: 7;
wherein (a), (b) and (c) are joined 5’-3’ in this order, and wherein said nucleic acid molecule is capable of promoting transcription of an operably-linked heterologous polynucleotide in a mammalian cell.
In some preferred embodiments, the nucleic acid molecule comprises:
(a) a first polynucleotide having the nucleotide sequence set forth in SEG ID NO: 1;
(b) a second polynucleotide having the nucleotide sequence set forth in SEQ ID NO: 4; and (c) a third polynucleotide having the nucleotide sequence set forth in SEG ID NO: 5;
wherein (a), (b) and (c) are contiguously joined 5’-3’ in this order, and wherein said nucleic acid molecule is capable of promoting transcription of an operably-linked heterologous polynucleotide in a mammalian ceil.
In some preferred embodiments, the nucleic acid molecule comprises:
(a) a first polynucleotide having the nucleotide sequence set forth in SEG ID NO: 2;
(b) a second polynucleotide having the nucleotide sequence set forth in SEG ID NO: 4; and (c) a third polynucleotide having the nucleotide sequence set forth in SEG ID NO: 6;
wherein (a), (b) and (c) are contiguously joined 5’-3’ in this order, and wherein said nucleic acid molecule is capable of promoting transcription of an operably-linked heterologous polynucleotide in a mammalian cell.
In some preferred embodiments, the nucleic acid molecule comprises:
(a) a first polynucleotide having the nucleotide sequence set forth in SEQ ID NO: 3;
(b) a second polynucleotide having the nucleotide sequence set forth in SEG ID NO: 4; and (c) a third polynucleotide having the nucleotide sequence set forth in SEG ID NO: 7;
wherein (a), (b) and (c) are contiguously joined 5’-3’ in this order, and wherein said nucleic add molecule is capable of promoting transcription of an operably-linked heterologous polynucleotide in a mammalian ceil.
As used herein, the term “heterologous polynucleotide” refers to a polynucleotide which encodes a desired mRNA or polypeptide. Examples of heterologous polynucleotides include those that encode polypeptides, e.g. viral or mammalian polypeptides.
In some preferred embodiments, the heterologous polynucleotide encodes a protein that enters the secretory pathway, e.g. a membrane-linked protein, a secreted antibody or other secreted protein. Cells expressing such heterologous polynucleotides would be particularly suited to continuous production of recombinant secreted proteins in a bioreactor or suspension culture vessel, where the product can be harvested from the culture medium without lysing the cells.
Other preferred heterologous polynucleotides are those that encode proteins that remain cell-associated, such as membrane proteins or cytoplasmic proteins, which can either be used in the context of the whole ceil or harvested by cell lysis. In some embodiments, the heterologous polynucleotide encodes an antibody. In some other preferred embodiments of the invention, the heterologous polynucleotide codes for a viral polypeptide. Preferably, the viral polypeptide is a surface glycoprotein, e.g. VSV G. The VSV G polypeptide is a single pass membrane glycoprotein derived from the Vesicular Stomatitis virus. It mediates a broad infectious tropism. In other embodiments, the viral polypeptide is Gag-Poi, Rev or Tat. The term “Gag-Pol” refers to a retrovirus protein that is proteolyticaily cleaved to produce a functional reverse transcriptase, integrase, and protease and at least two proteins of structural importance for virus assembly. Preferably, the Gag-Pol sequence is from a lentivirus, most preferably from HIV. The Rev protein aids transport of virus genomes into the cytoplasm. Preferably, the Rev polypeptide sequence is from a lentivirus, most preferably from HIV. The Tat protein enhances the efficiency of viral transcription. Preferably, the Tat polypeptide sequence is from a lentivirus, most preferably from HIV.
The first, second, third (when present) and heterologous polynucleotides (when present) are joined in this order in a 5’-3’ direction.
- 10One or more linker nucleotides may be present in between the first and second, second and third, second and heterologous, and third and heterologous polynucleotides, e.g, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 linker nucleotides. When present, such linker polynucleotides should not have a significant detrimental effect on the efficacy of the promoter fragment (as measured, for example, in a luciferase assay). Preferably, there are no linker nucleotides, i.e. preferably the first, second, third (when present) and heterologous polynucleotides (when present) are joined contiguously in this order in a 5’-3’ direction.
The nucleic acid molecule is capable of promoting transcription of an operablylinked heterologous polynucleotide in a mammalian ceil.
The heterologous polynucleotide will be operably placed at a position downstream (i.e. 3’) to the first, second and third (when present) polynucleotides of the invention.
In the context of the present invention, the term promoting transcription is intended to mean that when a heterologous polynucleotide is operably attached or inserted downstream of the first, second and third (when present) polynucleotides of the present invention, an expression product of the heterologous polynucleotide is obtained.
In some embodiments of the invention, the nucleic acid molecule is capable of promoting transcription of an operably-linked heterologous polypeptide in a mammalian cell in an inducible manner, i.e. the nucleic acid molecule is an inducible promoter. The nucleic acid molecule may therefore comprise an element which is repressible or activatable. For example, versions of the promoters disclosed here could be modified to contain binding sites for a protein that can either repress or activate transcription, creating inducible forms, in one example of this, the promoters disclosed herein (preferably p565 or p565i) could be modified to contain between 2-7 binding sites for the tetracycline repressor protein, most preferably 2-3 sites. By inserting said binding sites, the tetracycline repressor protein will be enabled to bind to the promoter and in doing so prevent the assembly of the basal transcription factor machinery on the promoter and thereby prevent both transcription and translation. This would abrogate protein expression. In the presence of Doxycycline or Tetracycline, the Tetracycline repressor protein is no longer able to bind DNA, and therefore the repressor can no longer bind the promoters, and transcription and translation can proceed unencumbered. As such,
- 11 the transcriptional activity of the promoter created can be said to be induced by the presence of either Doxycycline or Tetracycline and would therefore be classed as an inducible promoter.
The term expression product as used herein is intended to mean either or both of (i) RNA (e.g. hnRNA, mRNA, siRNA or miRNA) which is a transcribed product of the heterologous polynucleotide and (ii) a polypeptide which is a translated product of the heterologous polynucleotide.
The heterologous polynucleotide may be operably inserted downstream of the nucleic acid molecule of the present invention such that the 5'-terminai end of the heterologous polynucleotide is located in a region within 500 bp, 400 bp, 300 bp, 200 bp, 100 bp, 50 bp, 30 bp or 10 bp from the 3'-termina! end of the nucleic acid molecule of the present invention.
The level of transcription which has been promoted by the nucleic acid molecule of the invention may be assayed by any suitable method. For example, for such an assay, the heterologous polynucleotide may be a selection marker gene (e.g. a neomycin resistance gene or hygromycin B phosphotransferase gene) or an expression reporter gene (e.g. LacZ, GFP (Green Fluorescence Protein), luciferase genes, etc.). Preferably, confirmation of promoter activity may be accomplished by using a FLuc gene.
The transcriptional activity of the nucleic acid molecule of the invention may be measured by operably inserting a reporter gene, such as the FLuc gene, downstream of the nucleic acid molecule of the invention. The level of expression of firefly luciferase protein can then be used to indicate the level of transcription which is obtained. To achieve this, the required DNA (including the nucleic acid of the invention upstream of the Flue gene) may be introduced into a plasmid which is then transfected into suitable recipient cells (e.g. 293A cells) and allowed to express the firefly luciferase protein.
After 24 hours, the cells may be lysed and the luciferase in the cell lysate may be monitored using a luminometer by measuring its output of light in the presence of its luciferin substrate.
The nucleic acid molecule is capable of promoting transcription of an operablylinked heterologous polynucleotide in a mammalian cell. Preferred mammalian ceils include mouse, rat, hamster, monkey and human cells. Examples of such cells include
- 12HEK cells and derivatives (e.g. HEK293, HEK293T, HEK293A), PerC6, 911, CHO, HCT116, HeLa, COS and VERO ceils; cancer cells such as HepG2, A549, and MCF7; primary cells isolated from human or animal biopsies; and stem cells (including pluripotent cells such as embryonic stem cells and induced pluripotent stem (IPS) cells, as well as multipotent stem cells such as haematopoietic stem cells, mesenchymal stem cells, etc.).
Preferred human cells include HEK293, HEK293T and HEK293A cells; and human stem cells (including pluripotent cells such as embryonic stem cells and induced pluripotent stem (IPS) cells, as well as multipotent stem cells such as haematopoietic stem cells, mesenchymal stem ceils, etc.).
In a further embodiment, the invention provides an expression vector comprising a nucleic acid molecule of the invention. Preferably, the expression vector is a plasmid or virus vector. Examples of mammalian expression vectors include the adenoviral vectors, the pSV and the pCMV series of plasmid vectors, vaccinia and retroviral vectors, as well as baculovirus. in some embodiments, the expression vector is a ientivirai vector.
The expression vector may additionally comprise one or more of the following: an origin of replication, a selectable marker, and a multiple cloning site.
In yet a further embodiment, the invention provides an expression vector comprising the nucleic acid molecule of the invention, wherein the nucleic acid molecule is operably-linked to a heterologous polynucleotide.
The invention also provides a mammalian host ceil comprising an expression vector of the invention. The expression vector may be transfected into a host cell by any suitable method. Preferably, the host cell is a mammalian cell (e.g. a human cell), such as those mentioned above. Such host cells may be isolated ceils.
The invention further provides a mammal whose genome comprises a nucleic acid molecule of the invention or an expression vector of the invention. Preferably, the nucleic acid molecule of the invention or an expression vector of the invention is inserted into the genome of the mammal in such a way that a heterologous polynucleotide which is operably linked to said nucleic acid molecule of the invention or which is operably inserted into said expression vector of the invention is expressed in
- 13 one or more cells of the mammal. Preferably, the mammal Is a mouse or rat. In some embodiments, the mammal Is a non-human mammal.
The Invention also provides a kit comprising an expression vector and/or host cell of the Invention, optionally together with one or more additional components selected from the group consisting of:
(I) a helper plasmid (e.g. one containing a nucleotide sequence encoding a lentiviral polypeptide under regulatory control of a promoter of the invention);
(II) a virus genome plasmid (e.g. one with a packaging signal which may be adapted for simple insertion of a required transgene);
(iii) a buffer solution;
(iv) a restriction enzyme;
(v) transfection media; and (vi) mammalian cells.
Percentage amino acid sequence identities and nucleotide sequence identifies may be obtained using the BLAST methods of alignment (Alfschul et al. (1997),
Gapped BLAST and PSi-BLAST: a new generation of protein database search programs, Nucleic Acids Res. 25:3389-3402; and http://www.ncbi.nlm.nih.gov/BLAST). Preferably the standard or default alignment parameters are used.
Standard protein-protein BLAST (blastp) may be used for finding similar sequences in protein databases. Like other BLAST programs, blastp is designed to find local regions of similarity. When sequence similarity spans the whole sequence, blastp will also report a global alignment, which is the preferred result for protein identification purposes. Preferably the standard or default alignment parameters are used. In some instances, the low complexity filter may be taken off.
BLAST protein searches may also be performed with the BLASTX program, score-50, wordlength-S. To obtain gapped alignments for comparison purposes, Gapped BLAST (in BLAST 2.0) can be utilized as described in Altschul et al. (1997) Nucleic Acids Res. 25: 3389. Alternatively, PSI-BLAST (in BLAST 2.0) can be used to perform an iterated search that detects distant relationships between molecules. (See Altschul eta!. (1997) supra). When utilizing BLAST, Gapped BLAST, PSi-BLAST, the default parameters of the respective programs may be used.
- 14With regard to nucleotide sequence comparisons, MEGABLAST, discontiguousmegablast, and blastn may be used to accomplish this goal. Preferably the standard or default alignment parameters are used. MEGABLAST is specifically designed to efficiently find long alignments between very similar sequences. Discontiguous MEGABLAST may be used to find nucleotide sequences which are similar, but not identical, to the nucleic acids of the invention.
The BLAST nucleotide algorithm finds similar sequences by breaking the query into short subsequences called words. The program identifies the exact matches to the query words first (word hits). The BLAST program then extends these word hits in multiple steps to generate the final gapped alignments. In some embodiments, the BLAST nucleotide searches can be performed with the BLASTN program, score=100, wordlength=12.
One of the important parameters governing the sensitivity of BLAST searches is the word size. The most important reason that blastn is more sensitive than MEGABLAST is that it uses a shorter default word size (11). Because of this, blastn is better than MEGABLAST at finding alignments to related nucleotide sequences from other organisms. The word size is adjustable in blastn and can be reduced from the default value to a minimum of 7 to increase search sensitivity.
A more sensitive search can be achieved by using the newly-introduced discontiguous megablast page (www.ncbi.nlm.nih.gov/Web/Newsitr/FailWinterO2/biastlab.html). This page uses an algorithm which is similar to that reported by Ma et al. (Bioinformatics. 2002 Mar; 18(3): 440-5). Rather than requiring exact word matches as seeds for alignment extension, discontiguous megablast uses non-contiguous word within a longer window of template. In coding mode, the third base wobbling is taken into consideration by focusing on finding matches at the first and second codon positions while ignoring the mismatches in the third position. Searching in discontiguous MEGABLAST using the same word size is more sensitive and efficient than standard blastn using the same word size. Parameters unique for discontiguous megablast are:
word size: 11 or 12; template: 16, 18, or 21; template type: coding (0), non-coding (1), or both (2).
- 15 BRIEF DESCRIPTION OF THE FIGURES
Figure 1: pSF-SnapFast FLuc reporter vector background map.
Figure 2: Chart comparing recombinant promoter fragments with a range of standard promoters. Error bars show standard error. The experiment was performed in triplicate.
Figure 3: Graph comparing the highest performing three recombinant promoters (p585, p587 and p576) with a range of standard high-expression promoters in a time course assay.
Figure 4: Protein expression levels driven by recombinant promoter p585.
Figure 5: Protein over expression in HEK293 cells from the CMV promoter and
Oxford Genetics ‘hybrid’ prom-565 promoter using plasmid vector pcDNA3.1 and SnapFast Prolv, respectively. (pcDNA3.1 is a protein expression vector available from Life Technologies. The plasmid vector contains the immediate early CMV promoter, the T7 promoter, the Bovine Growth Factor (BgH) poly adenylafion signal alongside the F1 origin for making single stranded DNA and the SV40 promoter driving the expression of neomycin-kanamycin phosphotransferase (Aminoglycoside-3'-phosphotransferase) and the SV40 poly adenylafion signal. If the pcDNA3.1 vector is to be used to express an exogenous protein of interest, the coding sequence of said protein would need to be inserted downstream (3’) to the CMV and T7 promoters but upstream (5’) of the BgH polyadenylation signal in the 5’ to 3’ orientation. The plasmid also contains an Ampicillin resistance gene for selection in bacterial cells. SnapFast ProV1 is a protein expression vector designed by Oxford Genetics Ltd that contains the promoter termed herein p565i followed by the SV40 polyadenylation signal, if the SnapFast ProV1 vector is to be used to express an exogenous protein of interest, the coding sequence of said protein would need to be inserted downstream (3’) to the p565i promoter but upstream (5’) of the SV40 polyadenylation signal in the 5’ to 3’ orientation. The plasmid also contains a Kanamycin resistance gene for selection in bacterial ceils.) HEK293 cells were transfected with either pcDNA3.1 or SnapFast Prolv expressing various FLAG-tagged fusion proteins. Protein levels from culture supernatant or ceil lysate were determined by Western blot 72 hours post-transfection using Protein Simple Wes™ automated Western blot system. The FLAG-tagged fusion proteins were detected with mouse antiFLAG primary antibody and secondary rabbit anti-mouse-IgG-HRP.
- 16Figure 6: Protein over expression in HEK293 cells from the CMV promoter and Oxford Genetics ‘hybrid’ prom-565 promoter using plasmid vector pcDNA3.1 and SnapFast Prolv, respectively. HEK293 cells were transfected with either pcDNA3.1 or SnapFast Prolv expressing various FLAG-tagged fusion proteins. Protein levels from culture supernatant or cell lysate were determined by Western blot 72 hours posttransfection using Protein Simple Wes™ automated Western blot system. The FLAGtagged fusion proteins were detected with mouse anti-FLAG primary antibody and secondary rabbit anti-mouse-IgG-HRP.
Figure 7: Protein over expression in Chinese hamster ovary (CHO) ceils from the CMV promoter and Oxford Genetics ‘hybrid’ prom-565 promoter using plasmid vector pcDNA3.1 and SnapFast Prolv, respectively. CHO cells were transfected with either pcDNA3.1 or SnapFast Prolv expressing various FLAG-tagged fusion proteins. Protein levels from culture supernatant or cell lysate were determined by Western blot 72 hours post-transfection using Protein Simple Wes™ automated Western blot system. The FLAG-tagged fusion proteins were detected with mouse anti-FLAG primary antibody and secondary rabbit anti-mouse-IgG-HRP.
Figure 8: Protein over expression in Chinese hamster ovary (CHO) ceils from the CMV promoter and Oxford Genetics ‘hybrid’ prom-565 promoter using plasmid vector pcDNA3.1 and SnapFast Prolv, respectively. CHO cells were transfected with either pcDNA3.1 or SnapFast Prolv expressing various FLAG-tagged fusion proteins. Protein levels from culture supernatant or cell lysate were determined by Western blot 72 hours post-transfection using Protein Simple Wes™ automated Western blot system. The FLAG-tagged fusion proteins were detected with mouse anti-FLAG primary antibody and secondary rabbit anti-mouse-IgG-HRP.
Figure 9: Expression levels of GFP driven by promoters of the invention in HCT116 cells.
Figure 10: Expression levels of GFP driven by promoters of the invention in A549 ceils.
- 17ΕΧΑΜ PILES
The present invention is further illustrated by the following Examples, in which parts and percentages are by weight and degrees are Celsius, unless otherwise stated. It should be understood that these Examples, while indicating preferred embodiments of the invention, are given by way of illustration only. From the above discussion and these Examples, one skilled in the art can ascertain the essential characteristics of this invention, and without departing from the spirit and scope thereof, can make various changes and modifications of the invention to adapt it to various usages and conditions. Thus, various modifications of the invention in addition to those shown and described herein will be apparent to those skilled in the art from the foregoing description. Such modifications are also intended to fail within the scope of the appended claims.
The disclosure of each reference set forth herein is incorporated herein by reference in its entirety.
Example 1: Recombinant promoter fragments
Recombinant promoter fragments were produced consisting of the sequences identified in Table 1.
Table 1: Sequences of recombinant promoter fragments
Promoter Name First Polynucleotide Second Polynucleotide Third Polynucleotide
p567 SEQ ID NO: 3 SEQ ID NO: 4
p567i SEQ ID NO: 3 SEQ ID NO: 4 SEQ ID NO: 7
p576 SEQ ID NO: 2 SEQ ID NO: 4
p576i SEQ ID NO: 2 SEQ ID NO: 4 SEQ ID NO: 6
p565 SEQ ID NO: 1 SEQ ID NO: 4
p565i SEQ ID NO: 1 SEQ ID NO: 4 SEQ ID NO: 5
The first, second and third (when present) polynucleotides were joined contiguously in the above promoter fragments.
- 18Example 2: Construction of expression vector comprising promoter The reporter vector that was used in the Firefly Luciferase assays was constructed as follows.
Preparing the vector
The SnapFast (SF) vector shown in Figure 1 was used. The back-bone of the SnapFast vector consists of a pUC bacterial origin of replication, an ampicillin resistance gene and a multiple cloning site. A firefly luciferase coding sequence was cloned into the multiple cloning site between the Ncol and Xbal restriction sites (see Figure 1). The luciferase coding sequence incorporates a Kozak ribosome binding site.
The reporter plasmid was linearized with Bglll and then de-phosphorylated to prevent re-ligation of the vector backbone. The cut vector was isolated from uncut vector by gel extraction followed by column based purification. The recombinant promoter fragments (from Example 1) were then ligated info the Bglll cut vector.
The vectors were transformed into a standard E co// cloning strain and plated onto LB kanamycin selection plates resulting in one recombinant promoter per colony. Colonies were picked for mini scale-plasmid preparation. These promoter clones were used for transfections into HEK293 cells and subsequent luciferase expression assays.
Example 3: Assay for promoter activity
Materials and Methods
Plasmid DNA was purified from a colony from each recombinant promoter. The reporter vectors containing the recombinant promoters were individually transfected into HEK293 cells in several 96~well plates. The transfected cells were incubated at 37°C for 24 hours. Each well was assayed for luciferase activity using the Luciferase Assay System kit (Promega, Wisconsin, USA).
Briefly, each assay was performed as follows: Culture medium was removed from each well. 100 μΙ of reporter lysis buffer was added to the cells in each well. The microtitre plate(s) was the incubated for 30 minutes at -20°C to lyse the cells, then the plate was thawed by incubating it for 30 minutes at room temperature. The ceil lysates were homogenised by pipetting up and down. A 25 μΙ sample of each lysate was
- 19transferred into a luminometer tube. The Iuciferase assay reagent was reconstituted by adding iuciferase assay buffer to it. Samples were analysed on a Lumat LB 9507 luminometer (EG&G Berthold). The luminometer was set to inject 25 pi of Iuciferase assay reagent and then record the light emitted for a duration of 2 seconds. The output of the luminometer was given in relative light units.
The recombinant promoters were compared against a selection of standard highly-expressing promoters.
Results
The results are shown in Figure 2. The recombinant promoter fragments p567 and p576 performed better than CMV (the current gold standard) in this assay.
Example 4: Time course experiment
Three recombinant promoters (p565, p567 and p576) were selected for a timecourse experiment. The results are shown in Figure 3.
The results show that the recombinant promoters gave rise to consistently high Iuciferase expression over the course of 24 hours to 72 hours post-transfection.
Example 5: Expression levels from the recombinant promoters
Protein expression levels driven by recombinant promoter p565 of the invention are shown in Figure 4, with CMV promoter expression shown as a control. The p565 promoter was compared with a standard CMV promoter for expression of 4 different commercially useful antigens (Proteins ‘W’, ‘X’, Ύ and !Z’). For each antigen, the coding sequence was cloned downstream of either the CMV or p565 promoter in a vector equivalent to the one shown in Figure 1.
On day 1, a 48-weli microtiter plate was seeded with HEK293Ad ceils at a density of 30,000 ceils per well in a volume of 300 pi of serum free culture medium. On day 2, the ceils in each well were transfected with 0.75 pg of DNA for each of the expression vectors. The cells were transfected using branched polyethylenimine (PEI) at a ratio of 3 pg for every pg of DNA. Prior to adding the DNA to the cells, it was incubated with the PEI for 20 minutes at room temperature to form DNA:PEI complexes.
-20Three days after the transfection took place, the supernatant from each well was harvested and analysed for the quantity of antigen expressed by Western blot.
Example 6: Expression levels in HEK293 cells
Various human genes (see Table 2) were over-expressed from the CMV promoter or promoter P565I in human embryonic kidney cells (HEK293) by a method of transient transfection using branched PEI (25kDa).
Table 2: Human genes which were overexpressed in HEK293 cells
Ptoisir Rsf
KIF4 >sp I O43474iKl.F4_RUMAK Krueppel-iike fector 4
XiAP >spi Ρ3817Ο|Χ!ΑΡ.HUMAN £3 ubiquitin-protein iigase XiAP
CYP2C1 >spi P3326L NP2Ci_ BISMAKi Cytochrome P4SQ2QS
CE54 >sp! Pi6OTOjCD44_HU?i4AN CB44antfgen
Myc >sp: ^01.3.06 SViyc proto-oncogene protein
LOX >spi P283«5iLYOX_HUMAN Protetn^ysine δ-oxidase
RAk! >sp: 2 826 j8AGMAh; GTP-hindsng niitdesr protect Rar:
FEV »sp|Q9958iiFEV_HUMAN Protein FEV
ABO >sp ί P164A2 S8GA7_HU?iWi Hisfo-biosxi group ABO system transferase
GAP'D!-! >· <: PG44D6 ^33P_HL? Giyter aidehyde-S-phospEtata dehydrogenase
CDK >spi P064S3iCDKi_HUMAN Cydin-dependent kinase I
>sp: Q99836j MYE>88_HUMAN Myektid differentiation primary response protein
CRP >spi PO2741 |CRS>_HLIMA8 C-reactive protein
NBAS POilll -nASts HUMAG GTPase ?A(,<·
Protein Ref
SLiO >sp|P223Ql iiLiO_HUMAN i«terfeukto-IQ
ΡΪΗ >sp | PO127O | Parathyroid hormone
TNF >sp j POI375 JTff Τοπ,οτ necrosis factor
SMS >?.[-! jPOl TOR jiMS_HUMAN insuSn
CM :--s p f POO915 (CAHi_K U MAN Carbon ic a n h yd rase
JUki >sp IPC'5412 iJUNJftjMANTranscription faster AP-i
KRAS >sp {POI1161 RASX_HUMAN GTFase KSas
CSSS >sp IP348K5 iCBSSHUMAN Masrosiaiin
CD14 >sp|PO85?i SCBMWfMAN Monocyte differentiation antigen CD 14
CD34 >sp|P28906iCB34 HUMA8 Hematopoietic progenitor cett 3ntigen CB34
PhcsphaiidySmosftoi 3A>-iFisphosphat& 3-phosphatase disahspsctfiaty protein phosphatase PTE^
T?53 >sp|P04&37 iPS3_HUMANCellolar tumorantigee p53
NANOG >sp!QSHSSOf NANG6_HUMAN Homeobar protein NASiOG
RPE > | Q96ATS (RPE_HUMAN Ribuiose-phosphste 3-epifnerase
HEK293 cells were seeded In tissue culture In a 48-wel! plate format (Sigma Aldrich) at a density of 25,000 cells per well for 24-hours prior to plasmid DNA transfection. Each plasmid DNA (750 ng) expressing FLAG tagged human gene from the CMV promoter or promoter P565I was mixed with branched PEI (25kDA) at a ratio of 1:3, and the DNA:PEI complex were transiently transfected into each well of 48-weil plate seeded HEK293 cells. Expression of each FLAG tagged human protein from transient transfection was analysed by automated western blotting (Wes™ Protein
Simple) 72-hours post-transfection using mouse anti-FLAG primary antibody and secondary rabbit anti-mouse-IgG-HRP. The results are shown in Figures 5 and 6.
Example 7: Expression levels in CHO ceils
Various human genes (see Table 3) were over-expressed from the CMV promoter or promoter P565I in CHO K1 cells by a method of transient transfection using branched PEI (25kDa).
Table 3: Human qenes which were overexpressed in CHQ cells
SWA 8«f
8A$
W A«ASS AfessEEV
au? w!!A2 feCm-AWo»;.',y:a;-?iks 2
CAS VfpsPGEiSSSSkAHS^HtfA^ESsfefSk'AvtikSSSS
xx! Xi.iph^SS'^s^T.m.FSFSE.'W AfefegesicsciiEswAA' -pmehs, rmvEoAfe
W ΐ GfeS A J&MASS WAiS AvWki A prtsriirnf $WO«$e proto»
S3fJ Wi PSWSW U»Wf W A-A growfe few S
SAv w| iAogsW
FSXS w 1 F'feife? 1 EKH»«8 SA
wu s*p ϊ RW1Ϊ fern Ji S&SAA SAAwAA 9? A sw A
ms >s:^ j A jfes A ifGfeVf W A
w J8»jTO® jRMSLi««Mi Astfef «Site
Proisifi Ref
CRP >sp f PO274S |CRP_HUMAM C-wsacfive protein
TSE >sp ί FOI375 p‘!\FA__H’JMAisi Τυη:υ< ^ecfosisfeetOF
BFiDF >| P23S6O| BB?iF_HUMAhi Brain-derived neurotrophic factor
FGF23 >sp iG9SZV9 iFSF23 .HUMAN Fibrobiast growth factor 23
APOE >sp1P02649 j APOE_HUMAfi Apolipoprotein
CCLZ >sp j PS35QOICCL2HUMAN C-C motif chemoidne 2
VAX >sp | F61244]MKX_HOMAN Protein max
FRi'S >sp f POill.6 jRASKHUMAif STPase XRas
XiRAS ash j P01 Ill jRASNHUMAti STPase NRas
BAX >sp f Q078121BAXHUMAN Apoptosis regulator BAX
RACS sspiPSSOOO ACl.HLiMA!<i Ras-reiated C3 footulinum toxin substrate i
RHCSA >sp (P6S586 |RHOA_HUMAHTransforming protein RhoA
EPO >sp (POiS88|EPO_HUMAN Erythropoietin
HMGB1 >«p|PGS429jBMGBl. HUMAN High mobility group protein BI
-23 CHO K1 ceils were seeded in tissue culture in a 48-weil plate format (Sigma Aldrich) at a density of 20,000 ceils per well for 24-hours prior to plasmid DNA transfection. Each plasmid DNA (750 ng) expressing FLAG tagged human gene from the CIVSV promoter or promoter P565I was mixed with branched PEI (25kDA) at a ratio of 1:3, and DNA:PEI complex were transiently transfected into each well of 48-weil plate seeded CHO K1 ceils. Expression of each FLAG tagged human protein from transient transfection was analysed by automated western blotting (Wes™ Protein Simple) 72hours post-transfection using mouse anti-FLAG primary antibody and secondary rabbit anti-mouse-IgG-HRP. The results are shown In Figures 7 and 8.
Example 8: Expression levels in HCT116 ceils
Green fluorescent protein (GFP) was expressed from the CMV, P565i, P565, P576, P567 promoters in HCT116 cells by a method of transient transfection using branched PEI (25kDa).
HCT116 ceils were seeded in tissue culture treated 48-well plate format (Sigma Aldrich) at a density of 25,000 cells per well for 24-hours prior to plasmid DNA transfection. Each plasmid DNA (750 ng) expressing the enhanced green fluorescent protein from the CMV, P565I, P565, P576, or P567 promoter was mixed with branched PEI (25kDA) at a ratio of 1:3, and the DNA:PEI complexes were transiently transfected into HCT116 cells. Enhanced green protein expression as determined by MFI of HCT116 cells was measured by flow cytometry 48-hours post-transfection. The results are shown in Figure 9.
Example 9: Expression levels In A549 cells
Green fluorescent protein (GFP) was expressed from the CMV, P565i, P565, P576, P567 promoters in A549 cells by a method of transient transfection using branched PEI (25kDa).
A549 cells were seeded in tissue culture treated 48-well plate format (Sigma Aldrich) at a density of 25,000 cells per well for 24-hours prior to plasmid DNA transfection. Each plasmid DNA (750 ng) expressing the enhanced green fluorescent protein from the CMV, P565i, P565, P576, or P567 promoter was mixed with branched
-24PEi (25kDA) at a ratio of 1:3, and the DNA:PEI complexes were transiently transfected into A549 cells. Enhanced green protein expression as determined by MF! of A549 cells was measured by flow cytometry 48-hours post-transfection. The results are shown in Figure 10.
SEQUENCES
SEQ ID NO: 3
AGTAATCAATTACGGGGTCATTAGTTCATAGCCCATATATGGAGTTCCGCGTTACAT
AACTTACGGTAAATGGCCCGCCTGGCTGACCGCCCAACGACCCCCGCCC
SEQ ID NO: 2
CTACCGGGTAGGGGAGGCGCTTTTCCCAAGGCAGTCTGGAGCATGCGCTTTAGCA
GCCCCGCTGGGCACTTGGCGCTACACAAGTGGCCTCTGGCCTCGCACACATTCCA
CATCCACCGGTAGGCGTCAATGGAAAGTCCCTATTGGCGTTACTATGGGAACATAC
GTCATT
SEQ ID NO: 1
CGTGAGGCTCCGGTGCCCGTCAGTGGGCAGAGCGCACATCGCCCACAGTCCCCG
AGAAGTTGGGGGGAGGGGTCGGCAATTGAACCGGTGCCTAGAGAAGGTGGCGCG
GGGTAAACTGGGAAAGTGATGTCGTGTACTGGCTCCGCCTTTTTCCCGAGGGTGG
GGGAGAACCGTATATAAGTGCACTAGTCGCCGTGAACGTCAATGGAAAGTCCCTA
TTGGCGTTACTATGGGAACATACGTCATTATTGACGTCAATGACGGTAAATGGCCC
GCCTGGCATTATGCCCAGTACATGACCTTATGGGACTTTCCTACTTGGCAGTACAT
CTACGTATTAGTCATCGCTATTACCATGCTGATGCGGTTTTGGCAGTACATCAATG
GGCGTGGATAGCGGTTTGACTCACGGGGATTTCCAAGTCTCCACCCC
SEQ ID NO: 4
ATTGACGTCAATGGGAGTTTGTTTTGGCACCAAAATCAACGGGACTTTCCAAAATG
TCGTAACAACTCCGCCCCATTGACGCAAATGGGCGGTAGGCGTGTACGGTGGGA
GGTCTATATAAGCAGAGCTGGTTTAGTGAACCGTCAGATC
-25 SEQ ID NO: 7
GTTGTTCGCTTTGATAAACTTCCAGGATTCGGAGACAGTATTGAAGCTCAGGTACA
GAAATAATTTCACCTTTGTTTCTCTTTCTATTCAGTGTGGCAGATCTGTAAACGTTCA
CTCTTCACTTAGAGACATCCTCAACCAAATCACCAAACCAA
SEQ ID NO: 6
GCCCAGGAAGTACACGAGAAGCTCCGAGGATTGGCTGAAGTCCAACGTCTCTGAT
TGCGGTGGCTCAGAGCACCCGTATCATTTTGGAGGTGAGTGGCTTTGGTTCCCGG
CTGAGGTGGAGTGGGCTGAGGACTAGACTGAGCCCTCGGACATGGAGGTGGGGA
TGGGGCAGACTCATCCCATTCTTGACCAAGCCCTTGTTCTGCTCCCTTCCCAGGCT CTGTGACTGGGGCAACCTGCAAGGAGCTGGCCAGCCAGCCTGACGTGGACGGCT TCCTTGTGGGTGGTGCTTCCCTCAAGCCCGAATTCGTGGACATCATCAACGCCAAA CAA
SEQ ID NO: 5
TGAAGTTGGTGGTGAGGCCCTGGGCAGGTTGGTATCAAGGTTACAAGACAGGTTT
AAGGAGACCAATAGAAACTGGGCATGTGGAGACAGAGAAGACTCTTGGGTTTCTG
ATAGGCACTGACTCTCTCTGCCTATTGGTCTATTTTCCCACCCTTAG
SEQUENCE LISTING FREE TEXT <223> Synthetic promoter element

Claims (15)

1. A nucleic acid molecule comprising:
(a) a first polynucleotide having at least 80% nucleotide sequence identity to the nucleotide sequence set forth in SEQ ID NO: 1, SEQ ID NO: 2 or SEQ ID NO: 3, or a functional fragment of that first polynucleotide; and (b) a second polynucleotide having at least 80% nucleotide sequence identity to the nucleotide sequence set forth in SEQ ID NO: 4, or a functional fragment of that second polynucleotide; wherein (a) and (b) are joined 5!-3’, and wherein said nucleic acid molecule is capable of promoting transcription of an operably-linked heterologous polynucleotide in a mammalian cell.
2. A nucleic acid molecule as claimed in claim 1, wherein the first polynucleotide has at least 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% nucleotide sequence identity to one of the nucleotide sequences set forth in SEQ ID NO: 1, SEQ ID NO: 2 or SEQ ID NO: 3.
3. A nucleic acid molecule as claimed in any one of the preceding claims, wherein the second polynucleotide has at least 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% nucleotide sequence identity to the polynucleotide sequence set forth in SEQ ID NO: 4.
4. A nucleic acid molecule as claimed in any one of the preceding claims, wherein the nucleic acid molecule comprises:
(a) a first polynucleotide having the nucleotide sequence set forth in SEQ ID NO:
1, SEQ ID NO: 2 or SEQ ID NO: 3; and (b) a second polynucleotide having the nucleotide sequence set forth in SEQ ID
NO: 4.
5. A nucleic add molecule as claimed in any one of the preceding claims, wherein the nucleic acid molecule additionally comprises:
-27(c) a third polynucleotide having at least 80% nucleotide sequence identity to the nucleotide sequence set forth in SEQ ID NO: 5, SEQ ID NO:
6 or SEQ ID NO: 7, or a functional fragment of that third polynucleotide;
wherein (a), (b) and (c) are joined 5’-3’ in this order, and wherein said nucleic acid molecule is capable of promoting transcription of an operably-linked heterologous polynucleotide in a mammalian cell.
8. A nucleic add molecule as claimed in claim 5, wherein the third polynucleotide has at least 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% nucleotide sequence identity to one of the nucleotide sequences set forth in SEQ ID NO: 5, SEQ ID NO: 6 or SEQ ID NO: 7.
7. A nucleic acid molecule as claimed in any one of the preceding claims, wherein the nucleic acid molecule comprises:
(a) a first polynucleotide having the nucleotide sequence set forth in SEQ ID NO: 1, SEQ ID NO: 2 or SEQ ID NO: 3;
(b) a second polynucleotide having the nucleotide sequence set forth in SEQ ID NO: 4; and (c) a third polynucleotide having the nucleotide sequence set forth in SEQ ID NO: 5, SEQ ID NO: 6 or SEQ ID NO: 7;
wherein (a), (b) and (c) are joined 5’~3’ in this order, and wherein said nucleic acid molecule is capable of promoting transcription of an operably-linked heterologous polynucleotide in a mammalian cell.
8. A nucleic acid molecule as claimed in any one of the preceding claims, wherein the nucleic acid molecule additionally comprises an inducible element, preferably a repressible element or an activatable element.
9. A nucleic acid molecule as claimed in any one of the preceding claims, wherein mammalian cell is a mouse, rat, hamster, monkey or human cell.
-2810. An expression vector comprising a nucleic acid molecule as claimed in any one of the preceding claims.
11. An expression vector as claimed in claim 10, wherein the expression vector is an 5 adenoviral vector, a pSV or pCMV plasmid vector, a vaccinia or retroviral vector, a baculovirus vector, preferably a lentiviral vector.
12. An expression vector as claimed in claim 10 or claim 11, wherein the nucleic acid molecule is operably-linked to a heterologous polynucleotide.
13. A mammalian host cell comprising an expression vector as claimed in any one of claims 10 to 12.
14. A mammal whose genome comprises a nucleic acid molecule as claimed in any 15 one of claims 1 -9 or an expression vector as claimed in any one of claims 10-12.
15. A kit comprising an expression vector as claimed in any one of claims 10-12 and/or a host cell as claimed in claim 13, optionally together with one or more additional components selected from the group consisting of:
20 (i) a helper plasmid:
(ii) a virus genome plasmid;
(iii) a buffer solution;
(iv) a restriction enzyme;
(v) transfection media; and (vi) mammalian cells.
Intellectual
Property
Office
Application No: Claims searched:
GB1703224.4A 2016-03-01 2017-02-28 Promoter Active GB2553170B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
GB2002193.7A GB2578859B (en) 2016-03-01 2017-02-28 Promoter

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
GBGB1603577.6A GB201603577D0 (en) 2016-03-01 2016-03-01 Promoter

Publications (3)

Publication Number Publication Date
GB201703224D0 GB201703224D0 (en) 2017-04-12
GB2553170A true GB2553170A (en) 2018-02-28
GB2553170B GB2553170B (en) 2020-05-06

Family

ID=55807149

Family Applications (3)

Application Number Title Priority Date Filing Date
GBGB1603577.6A Ceased GB201603577D0 (en) 2016-03-01 2016-03-01 Promoter
GB2002193.7A Active GB2578859B (en) 2016-03-01 2017-02-28 Promoter
GB1703224.4A Active GB2553170B (en) 2016-03-01 2017-02-28 Promoter

Family Applications Before (2)

Application Number Title Priority Date Filing Date
GBGB1603577.6A Ceased GB201603577D0 (en) 2016-03-01 2016-03-01 Promoter
GB2002193.7A Active GB2578859B (en) 2016-03-01 2017-02-28 Promoter

Country Status (9)

Country Link
US (1) US10647999B2 (en)
EP (2) EP3423583B1 (en)
JP (1) JP6960409B2 (en)
KR (1) KR20180132045A (en)
CN (1) CN109072251A (en)
AU (1) AU2017225350B2 (en)
GB (3) GB201603577D0 (en)
SG (1) SG11201806782QA (en)
WO (1) WO2017149292A1 (en)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110892064A (en) 2017-07-25 2020-03-17 牛津遗传学有限公司 Adenoviral vectors
CN107630035A (en) * 2017-09-28 2018-01-26 兰州大学 A kind of rat alexin carrier for expression of eukaryon and its construction method
GB201800903D0 (en) 2018-01-19 2018-03-07 Oxford Genetics Ltd Vectors
AU2019215152A1 (en) * 2018-02-01 2020-08-20 Homology Medicines, Inc. Adeno-associated virus compositions for restoring PAH gene function and methods of use thereof
TW202208632A (en) 2020-05-27 2022-03-01 美商同源醫藥公司 Adeno-associated virus compositions for restoring pah gene function and methods of use thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012112603A1 (en) * 2011-02-14 2012-08-23 University Of Utah Research Foundations Constructs and methods for generating induced pluri potent stem ( ips) cells
US20120244185A1 (en) * 2009-09-16 2012-09-27 Vaxart ,Inc. Immunization strategy to prevent h1n1 infection
US20130164326A1 (en) * 2006-02-28 2013-06-27 Vaxart, Inc. Chimeric adenoviral vectors
CN104630267A (en) * 2014-07-17 2015-05-20 清华大学 Kit for constructing synthetic gene circuits by utilizing modularization of TALE transcriptional suppressors in mammal animals
WO2016022075A1 (en) * 2014-08-08 2016-02-11 Agency For Science, Technology And Research Mutants of the bacteriophage lambda integrase

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007081336A1 (en) * 2006-01-13 2007-07-19 Five Prime Therapeutics, Inc. Mammalian vectors for high-level expression of recombinant proteins
WO2008051854A2 (en) * 2006-10-20 2008-05-02 Trustees Of Boston University A tunable genetic switch for regulating gene expression
WO2008073303A2 (en) * 2006-12-07 2008-06-19 Switchgear Genomics Transcriptional regulatory elements of biological pathways, tools, and methods
DK2611915T3 (en) * 2010-09-01 2015-06-22 Cellagenics B V Nucleic acid fragments from a ribosomal protein promoter to enhance gene expression
WO2012136788A1 (en) 2011-04-07 2012-10-11 Bayer Cropscience Nv Seed - specific promoter in cotton
EP2771465A1 (en) * 2011-10-28 2014-09-03 Arizona Board of Regents, a Body Corporate of the State of Arizona acting for and on behalf of Arizona State University Genetic element that enhances protein translation
PL2711426T3 (en) * 2012-09-24 2015-09-30 Lonza Biologics Plc Expression vectors comprising chimeric cytomegalovirus promoter and enhancer sequences
KR102365484B1 (en) 2013-03-12 2022-02-18 에이쥐씨 바이올로직스, 인크. Improved recombinant protein expression using a hybrid chef1 promoter

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130164326A1 (en) * 2006-02-28 2013-06-27 Vaxart, Inc. Chimeric adenoviral vectors
US20120244185A1 (en) * 2009-09-16 2012-09-27 Vaxart ,Inc. Immunization strategy to prevent h1n1 infection
WO2012112603A1 (en) * 2011-02-14 2012-08-23 University Of Utah Research Foundations Constructs and methods for generating induced pluri potent stem ( ips) cells
CN104630267A (en) * 2014-07-17 2015-05-20 清华大学 Kit for constructing synthetic gene circuits by utilizing modularization of TALE transcriptional suppressors in mammal animals
WO2016022075A1 (en) * 2014-08-08 2016-02-11 Agency For Science, Technology And Research Mutants of the bacteriophage lambda integrase

Also Published As

Publication number Publication date
GB201603577D0 (en) 2016-04-13
AU2017225350B2 (en) 2023-07-20
EP3423583A1 (en) 2019-01-09
KR20180132045A (en) 2018-12-11
AU2017225350A1 (en) 2018-09-06
GB2578859A (en) 2020-05-27
US10647999B2 (en) 2020-05-12
US20190071692A1 (en) 2019-03-07
WO2017149292A1 (en) 2017-09-08
JP2019506882A (en) 2019-03-14
SG11201806782QA (en) 2018-09-27
EP3686282A1 (en) 2020-07-29
GB2578859B (en) 2020-10-28
JP6960409B2 (en) 2021-11-05
EP3423583B1 (en) 2023-05-24
CN109072251A (en) 2018-12-21
GB201703224D0 (en) 2017-04-12
GB202002193D0 (en) 2020-04-01
GB2553170B (en) 2020-05-06

Similar Documents

Publication Publication Date Title
US10647999B2 (en) Promoter
Trepotec et al. Segmented poly (A) tails significantly reduce recombination of plasmid DNA without affecting mRNA translation efficiency or half-life
Park et al. A plant viral “reinitiation” factor interacts with the host translational machinery
Schambach et al. Improving transcriptional termination of self-inactivating gamma-retroviral and lentiviral vectors
Gamarnik et al. Interactions of viral protein 3CD and poly (rC) binding protein with the 5′ untranslated region of the poliovirus genome
Clever et al. RNA structure and packaging signals in the 5′ leader region of the human immunodeficiency virus type 1 genome
Herbreteau et al. HIV-2 genomic RNA contains a novel type of IRES located downstream of its initiation codon
Verma et al. Polypyrimidine tract-binding protein interacts with coxsackievirus B3 RNA and influences its translation
US11236363B2 (en) Hybrid proteins and uses thereof
Ly et al. Bipartite signal for genomic RNA dimerization in Moloney murine leukemia virus
Dalziel et al. Two G-rich regulatory elements located adjacent to and 440 nucleotides downstream of the core poly (A) site of the intronless melanocortin receptor 1 gene are critical for efficient 3′ end processing
CN107893085B (en) Dual-luciferase reporter gene plasmid for screening orphan receptor activity regulator and construction and application thereof
Chai et al. Human rhinovirus internal ribosome entry site element enhances transgene expression in transfected CHO-S cells
Ghaderi et al. Construction of an eGFP expression plasmid under control of T7 promoter and IRES sequence for assay of T7 RNA polymerase activity in mammalian cell lines
Chen et al. Functional characterization of a naturally occurring trans-splicing intein from Synechococcus elongatus in a mammalian cell system
WO2005090562A1 (en) Sequence capable of accelerating gene expression at moderately low temperature
Kamoshita et al. Translation initiation from the ribosomal A site or the P site, dependent on the conformation of RNA pseudoknot I in dicistrovirus RNAs
Ghassemi et al. Translational efficiency of BVDV IRES and EMCV IRES for T7 RNA polymerase driven cytoplasmic expression in mammalian cell lines
Tang et al. Alternative translation initiation of a haloarchaeal serine protease transcript containing two in-frame start codons
Yang et al. The 5′ untranslated region of the capsid protein 2 gene of mink enteritis virus is essential for its expression
Wei et al. Engineering artificial factors to specifically manipulate alternative splicing in human cells
Torkzaban et al. Tethered mRNA Amplifier: A Novel Approach to Increase Protein Expression
Manicone et al. Expression of HTLV-1 Genes in T-Cells Using RNA Electroporation
Bai et al. Prevalence of errors in lab-made plasmids across the globe
WO2017104796A1 (en) Artificial rna restriction enzyme